| Peer-Reviewed

The Use of a Shear Device to Monitor the Stability of a Single-Chain Variable Fragment (scFv) Fusion Protein MFECP1

Received: 17 March 2020    Accepted: 22 April 2020    Published: 29 May 2020
Views:       Downloads:
Abstract

Antibody based drugs are increasingly being used to treat a vast array of diseases because of their unique affinity to target specific antigen proteins on the surfaces of target cancer cells. Fusions of antibodies and conjugated biopharmaceuticals are progressively being used as this gives the opportunity to target other cytotoxic molecules to unwanted cells. It is critical to ensure these types of drug products are not fragile or uneconomical to produce at a large scale. A very small amount of precious protein solution can be characterised in an Ultra scale-down (USD) shear device to uncover if fusion proteins are prone to shear stress. This article presents how the purified and deglycosylated form of the MFECP1 fusion protein was quantified with an ELISA from 700-50 ng/ml, with a +/- 10% deviation in the standard curve. It also describes how the same MFECP1 fusion protein was analysed to establish the optimum experimental control conditions that were required to observe changes due to hydrodynamic-associated degradation in a shear device. Lastly, it looks at how a first order kinetic relationship can be used to model the rate of MFECP1 fusion protein degradation and how this was used to quantify the rate of protein loss during different shear environments with and without air/liquid interfaces.

Published in Bioprocess Engineering (Volume 4, Issue 1)

This article belongs to the Special Issue Advances in Biochemical Engineering and Biotechnology

DOI 10.11648/j.be.20200401.15
Page(s) 29-39
Creative Commons

This is an Open Access article, distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium or format, provided the original work is properly cited.

Copyright

Copyright © The Author(s), 2024. Published by Science Publishing Group

Keywords

Ultra Scale-down, Shear Device, Degradation, Fusion Proteins, USD

References
[1] Bagshawe, D. K. Antibody directed enzyme prodrug therapy (ADEPT). 1989, Br. Jr Cancer, 60, 275-281.
[2] Begent, R. H. J.; Verhaar, M. J.; Chester, K. A.; Casey, J. L.; Green, A. J.; Napier, M. P.; Hope-Stone, L. D.; Cushen, N.; Keep, P. A.; Johnson, C. J.; Hawkins, R. E.; Hilson, A. J. W.; Robson, L. 1996, Clinical evidence of efficient tumor targeting based on single-chain fv antibody selected from a combinatorial library. Nature Medicine, 9, 979-984.
[3] Bekard, B, I.; and Dunstan, E, D. (2009) Shear-Induced Deformation of Bovine Insulin in Couette Flow. J. Phys. Chem. B, 113, 8453–8457.
[4] Biddlecombe, G. J.; Craig, V. A; Zhang, H.; Uddin, S.; Mulot, S.; Fish, C. B.; Bracewell, G. D. 2007, Determining Antibody Stability: Creation of Solid-Liquid Interfacial Effects within a High Shear Environment. Biotechnology Progress 23, 1218–1222.
[5] Blas, P; Tolner, B; Ward, J; Chester, K; Hoare, M (2018) The Use of a Surface Active Agent in the Protection of a Fusion Protein during Bioprocessing. Biotechnol. Bioeng. 115, 11.
[6] Blas, P. 2019. Improving-the-Bioprocessing-of-ADEPT-Fusion-Proteins-using-Ultra-scale-down-techniques. Journal of bioengineering 1, 25-46.
[7] Boychyn, M.; Yim, S. S. S.; Shamlou, P. A.; Bulmer, M.; More, J.; Hoare, M. 2001, Characterization of flow intensity in continuous centrifuges for the development of laboratory equipment mimics, Chem. Eng. Sci. 56, 4759-4770.
[8] Bradford, (1976). A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal. Biochem. 72, 248–254.
[9] Charm, S. E.; Lai, C. J. 1971, Comparison of ultrafiltration systems for concentrations of biologicals. Biotechnol. Bioeng. 13, 185-202.
[10] Charm, S. E.; Wong, B. L. 1970, Enzyme inactivation with shearing. Biotechnol. Bioeng. 7, 1103-1109.
[11] Chattopadhyay, D.; Rathman, J. F.; Chalmers, J. J. 1994, The protective effect of specific medium additives with respect to bubble rupture. Biotechnol. Bioeng. 45, 473-480.
[12] Damodaran, S. 2003, In situ measurements of conformational changes in proteins at liquid interfaces by circular dichroism spectroscopy. Anal. Bioanal. Chem. 376, 182-188.
[13] Duerkop, M.; Berger, E.; Dürauer, A.; Jungbauer, A. (2018) Impact of Cavitation, High Shear Stress and Air/Liquid Interfaces on Protein Aggregation. Biotechnology Journal 13, 7.
[14] Goswami, S, Wang, W; Arakawa, T and Ohtake, S. 2013, Developments and Challenges for mAb-Based Therapeutics. Antibodies 2, 452-500.
[15] Harrison, J. S.; Gill, A.; Hoare, M. 1998, Stability of a single-chain fv antibody fragment when exposed to high shear environment combined with air-liquid interfaces. Biotechnol. Bioeng. 59, 517-519.
[16] Jeffrey, C. K.; Wei, S.; Priyanka, K.; Mostafa, K.; Xiaoli, W.; Yang, S.; Raimund, J. O.; and Ward, S. E. (2019) Engineering a HER2-specific antibody–drug conjugate to increase lysosomal delivery and therapeutic efficacy. Nature Biotechnology volume 37, 523–526.
[17] Lencki, R. W.; Tecante, A.; Choplin, C. 1993, Effect of shear on the inactivation kinetics of the enzyme dextransucrase. Biotechnol. Bioeng. 42, 1061-1067.
[18] Levy, M. S.; Collins, I. J.; Yim, S. S.; Ward, J. M.; Titchener-Hooker, N. J.; Shamlou, P. A.; Dunnill, P. 1999, Effect of shear on plasmid DNA in solution. Bioprocess Engineering, 20, 7-13.
[19] Maa, Y. F.; Hsu, C. C. 1996, Effect of high shear on proteins. Biotechnol. Bioeng 51, 458-465.
[20] Maa, Y. F.; Hsu, C. C. 1997, Protein denaturation by combined effect of shear and air-liquid interface. Biotechnol. Bioeng 54, 503-512.
[21] Medzihradszky, K, F.; Spencer, D, I.; Sharma, S, K.; Bhatia, J.; Pedley, R, B.; Read, D, A.; Begent, R, H.; Chester, K, A. (2004). Glycoforms obtained by expression in Pichia pastoris improve cancer targeting potential of a recombinant antibody-enzyme fusion protein. Glycobiology. 14, 27-37.
[22] Michael, P. N.; Chester, K. A.; Melton, R. G.; Robson, L.; Nicholas, W.; Boden, J. A.; Pedley, R. B.; Begent, R. H. J.; Sherwood, R. F.; Minton, N. P. 1996, ln vitro and in vivo characterisation of a recombinant carboxypeptidase G2: anti-CEA scFv fusion protein. Immuntechnology 2, 47-57.
[23] Oliva, A.; Santovena, A.; Farina, J.; Llabres, M. 2003, Effect of high shear rate on stability of proteins: Kinetinc study. J. Pharm. Biomed. Anal. 33, 145-155.
[24] Pedley, R. B,; Sharma, S. K.; Hawkins, R. E.; Chester, K. A. 2003, Antibody directed enzyme prodrug therapy. Method in molecular medicine. 90 491-515.
[25] Rayat, C, M, E.; Chatel, A.; Hoare, M.; Lye, J. G. 2016, Ultra scale-down approaches to enhance the creation of bioprocesses at scale: impacts of process shear stress and early recovery stages. Current Opinion in Chemical Engineering. 14, 150-157.
[26] Thomas, C. R.; Dunnill, P. 1979, Action of shear on enzymes: Studies with catalase and urease. Biotechnol. Bioeng. 11, 2279-2302.
[27] Thomas, R. C.; Geer, D. 2011, Effects of shear on proteins in solution. Biotechnology Letters 33, 443–456.
[28] Titchener-Hooker, N, J.; Dunnill, P.; Hoare, M (2008) Micro biochemical engineering to accelerate the design of industrial-scale downstream processes for biopharmaceutical proteins. Biotechnol. Bioeng, 100, 473–487.
[29] Tirrell, M.; Middleman, S.; Shear modification of enzyme kinetics. Biotechnol. Bioeng. 1975, 17, 299-303.
[30] Tolner, B.; Smith, L.; Begent, R. H. J.; Chester, K. A. 2006.(a) Production of recombinant protein in Pichia pastoris by Fermentation. Nature Protocols. 1, 1006-1021.
[31] Tolner, B.; Smith, L.; Begent, R. H. J.; Chester, K. A. 2006.(b) Expanded-bed adsorption immobilized-metal affinity chromatography. Nature Protocols. 1, 1213-1222.
[32] Vikar, P. D.; Narendranathan, T. J.; Hoare, M.; Dunnill, P. 1981, Studies of the effects of shear on globular proteins: Extentions to high shear fields and to pumps. Biotechnol. Bioeng. 23, 425-429.
[33] Yim, S, S.; Shamlou, P, A. (2000). The engineering effects of fluid flow on freely suspended biological macro-materials and macromolecules. Advances in Biochem Eng. 67, 83-121.
Cite This Article
  • APA Style

    Peter Blas. (2020). The Use of a Shear Device to Monitor the Stability of a Single-Chain Variable Fragment (scFv) Fusion Protein MFECP1. Bioprocess Engineering, 4(1), 29-39. https://doi.org/10.11648/j.be.20200401.15

    Copy | Download

    ACS Style

    Peter Blas. The Use of a Shear Device to Monitor the Stability of a Single-Chain Variable Fragment (scFv) Fusion Protein MFECP1. Bioprocess Eng. 2020, 4(1), 29-39. doi: 10.11648/j.be.20200401.15

    Copy | Download

    AMA Style

    Peter Blas. The Use of a Shear Device to Monitor the Stability of a Single-Chain Variable Fragment (scFv) Fusion Protein MFECP1. Bioprocess Eng. 2020;4(1):29-39. doi: 10.11648/j.be.20200401.15

    Copy | Download

  • @article{10.11648/j.be.20200401.15,
      author = {Peter Blas},
      title = {The Use of a Shear Device to Monitor the Stability of a Single-Chain Variable Fragment (scFv) Fusion Protein MFECP1},
      journal = {Bioprocess Engineering},
      volume = {4},
      number = {1},
      pages = {29-39},
      doi = {10.11648/j.be.20200401.15},
      url = {https://doi.org/10.11648/j.be.20200401.15},
      eprint = {https://article.sciencepublishinggroup.com/pdf/10.11648.j.be.20200401.15},
      abstract = {Antibody based drugs are increasingly being used to treat a vast array of diseases because of their unique affinity to target specific antigen proteins on the surfaces of target cancer cells. Fusions of antibodies and conjugated biopharmaceuticals are progressively being used as this gives the opportunity to target other cytotoxic molecules to unwanted cells. It is critical to ensure these types of drug products are not fragile or uneconomical to produce at a large scale. A very small amount of precious protein solution can be characterised in an Ultra scale-down (USD) shear device to uncover if fusion proteins are prone to shear stress. This article presents how the purified and deglycosylated form of the MFECP1 fusion protein was quantified with an ELISA from 700-50 ng/ml, with a +/- 10% deviation in the standard curve. It also describes how the same MFECP1 fusion protein was analysed to establish the optimum experimental control conditions that were required to observe changes due to hydrodynamic-associated degradation in a shear device. Lastly, it looks at how a first order kinetic relationship can be used to model the rate of MFECP1 fusion protein degradation and how this was used to quantify the rate of protein loss during different shear environments with and without air/liquid interfaces.},
     year = {2020}
    }
    

    Copy | Download

  • TY  - JOUR
    T1  - The Use of a Shear Device to Monitor the Stability of a Single-Chain Variable Fragment (scFv) Fusion Protein MFECP1
    AU  - Peter Blas
    Y1  - 2020/05/29
    PY  - 2020
    N1  - https://doi.org/10.11648/j.be.20200401.15
    DO  - 10.11648/j.be.20200401.15
    T2  - Bioprocess Engineering
    JF  - Bioprocess Engineering
    JO  - Bioprocess Engineering
    SP  - 29
    EP  - 39
    PB  - Science Publishing Group
    SN  - 2578-8701
    UR  - https://doi.org/10.11648/j.be.20200401.15
    AB  - Antibody based drugs are increasingly being used to treat a vast array of diseases because of their unique affinity to target specific antigen proteins on the surfaces of target cancer cells. Fusions of antibodies and conjugated biopharmaceuticals are progressively being used as this gives the opportunity to target other cytotoxic molecules to unwanted cells. It is critical to ensure these types of drug products are not fragile or uneconomical to produce at a large scale. A very small amount of precious protein solution can be characterised in an Ultra scale-down (USD) shear device to uncover if fusion proteins are prone to shear stress. This article presents how the purified and deglycosylated form of the MFECP1 fusion protein was quantified with an ELISA from 700-50 ng/ml, with a +/- 10% deviation in the standard curve. It also describes how the same MFECP1 fusion protein was analysed to establish the optimum experimental control conditions that were required to observe changes due to hydrodynamic-associated degradation in a shear device. Lastly, it looks at how a first order kinetic relationship can be used to model the rate of MFECP1 fusion protein degradation and how this was used to quantify the rate of protein loss during different shear environments with and without air/liquid interfaces.
    VL  - 4
    IS  - 1
    ER  - 

    Copy | Download

Author Information
  • The Kibworth School, Kibworth Beauchamp, United Kingdom

  • Sections